The similarities and differences between tumors can be shown by pan-cancer analysis, which can support to the theoretical framework guiding efforts in cancer prevention, the design of therapeutic targets, and the prospective screening of therapeutic drugs (38). SMAD3 is central to the TGF-β pathway and plays an important role in immune-inflammatory responses. Recent research suggests that SMAD3 is connected to the survival and prognosis of malignant tumors and plays a role in the etiology and advancement of colorectal cancer, lung cancer, breast cancer, bladder cancer, prostate cancer, cervical cancer, gastric cancer, and osteosarcoma(10, 11, 13, 39–42). We embarked on an exhaustive and all-encompassing analysis of 33 different global cancer types to discern the molecular characteristics of SMAD3. This analysis involved the utilization of diverse databases, including TCGA, GTEx, UALCAN, TIMER2.0, GSCA, and cBioportal. Our investigation aimed to elucidate SMAD3's role in tumor development and potential regulatory mechanisms, encompassing gene expression, prognosis, gene mutations, immunological infiltration, single-cell analysis, immune checkpoints, DNA methylation, RNA methylation, tumor mutation burden (TMB), microsatellite instability (MSI), tumor characteristics, and drug sensitivity.
Previous research has demonstrated that SMAD3 has a role in the progression and development of tumors and is expressed differently in cancer (43). Our findings demonstrate that SMAD3 is overexpressed in pancreatic cancer (PAAD) and adrenocortical cancer (ACC) compared to healthy tissues, consistent with prior investigations. Additionally, SMAD3 mRNA exhibited up-regulation in 13 other cancer types, with a down-regulation observed solely in 7 tumor types(BRCA, KIRC, OV, PRAD, SKCM, UCEC, and UCS). However, the data for SMAD3 expression differences in BLCA, CESC, KIRP, and READ were not statistically significant due to insufficient normal samples. The absence of normal samples also hindered the analysis of malignancies including DLBC, THYM, STES, UVM, PCPG, and MESO. Given the expanding dataset, this area warrants further investigation. Furthermore, our study revealed variable SMAD3 expression at different stages in PAAD, KIRC, BLCA, OV, KIPAN, BRCA, LUAD, and TGCT. We utilized immunohistochemistry (IHC) to validate the presence of SMAD3 protein in both cancer and normal tissue samples from PAAD, LIHC, GBM, and LUAD. In comparison to normal tissues, PAAD, LIHC, GBM, and LUAD exhibited higher levels of SMAD3 protein expression. Collectively, these findings suggest that SMAD3 may have a role in a variety of tumors and could serve as a prognostic predictor in pan-cancer.
Our Kaplan-Meier survival analysis, utilizing TCGA data, revealed that high SMAD3 expression was associated with poor prognosis in several cancer types, including LAML, PAAD, ACC, UVM, and LUAD. This finding aligns with earlier investigations, which have likewise reported a congruent predictive function for elevated SMAD3 expression in pancreatic ductal carcinoma and acute myeloid leukemia (44, 45). Conversely, elevated SMAD3 expression is linked to a favorable prognosis in patients with GBMLGG, KIRC, and KIPAN. Moreover, our study marked a pioneering investigation into the connection between SMAD3 promoter methylation and cancer. We uncovered a correlation between SMAD3 expression and DNA methylation, suggesting the potential utility of SMAD3 methylation level as a prognostic biomarker in cancer patients.
A genetic component contributing to a poor clinical outcome in LUAD and oral cancer is SMAD3 mutations (39, 46). We may conclude that SMAD3 is mutated in the majority of tumor types from the data analyzed in the cBioPortal platform. COAD, UCEC, and SKCM were the tumors with the highest incidence of SMAD3 variation, predominately mutation, followed byamplification. Moreover, SMAD3 expression correlates with CNV in GBMLGG, LGG, CESC, COAD, COADREAD, BRCA, STES, SARC, STAD, HNSC, LIHC, and SKCM. hence, we should focus on the association between SMAD3 gene mutation and these cancers.
An integral component of the tumor microenvironment (TME), tumor immunity is closely related to tumor immunity and drives tumor growth(47–49). The majority of TIME's components are immune cells(50, 51). Data extracted from the TIMER2.0 database unveiled a significant relationship between diverse immune cell infiltration and SMAD3 expression across various cancer types. Moreover, our analysis encompassed the assessment of immune and stromal components within the tumor microenvironment (TME), utilizing the immunological score and stromal score. Additionally, the pivotal parameter characterizing the status of the tumor microenvironment, specifically tumor purity, was indirectly assessed via the ESTIMATE score(52).
The findings indicate a robust positive association among the three immune infiltration scores in cancer types including LGG, COAD, COADREAD, PRAD, NB, READ, and UVM. In contrast, cancer types such as STES, KIRP, KIPAN, HNSC, KIRC, LUSC, LIHC, WT, BLCA, THCA, and ACC exhibited simultaneous negative associations. Furthermore, the study revealed a significantly positive association between immunomodulatory genes and SMAD3 expression in malignant tumors. These findings suggest that SMAD3 may contribute to the regulation of tumor immunity through its impact on immune infiltration.
Tumor mutation burden (TMB), microsatellite instability (MSI), and programmed death-ligand 1 (PD-L1) are frequently employed as markers to anticipate the efficacy of immune checkpoint inhibitors (ICIs). TMB, in particular, exhibits a strong correlation with the efficacy of PD-1/PD-L1 inhibitors, and the majority of clinical studies implementing TMB as a predictive marker have achieved their endpoints with remarkable success rates (53).
As a result, specific cancer patients may utilize the TMB marker to partially predict the efficacy of immunotherapy. Elevated TMB can also signify increased susceptibility of tumors to immune checkpoint inhibitors (ICIs). Notably, high SMAD3 expression demonstrated a positive correlation with enhanced TMB in GBMLGG, BRCA, LUAD, KIPAN, PRAD, KIRC, THCA, SKCM, BLCA, and DLBC, while it was associated with reduced TMB in SARC, HNSC, ACC, THYM, and PAAD. Moreover, microsatellite instability (MSI) pertains to the emergence of novel microsatellite alleles at a microsatellite locus within a tumor, in contrast to its normal tissue counterpart. MSI occurs due to insertions or deletions of duplicate units and is primarily attributable to functional flaws in DNA mismatch repair within tumor tissue (54). Mismatch repair (MMR) deficiencies can render tumors incapable of rectifying errors in DNA replication, leading to the accumulation of mutations and heightened microsatellite instability. The effectiveness of immune checkpoint inhibitors (ICIs) is directly proportional to the number of neoantigens recognized by immune cells. Furthermore, a positive correlation between SMAD3 and microsatellite instability (MSI) was observed in GBMLGG, COAD, SARC, SARC KIPAN, LUSC, and UVM. Additionally, we investigated the relationship between SMAD3 and tumor stemness. Cancer stem cells possess the unique ability to regenerate and generate a variety of tumor cells, making them indispensable in the contexts of tumor survival, growth, metastasis, and recurrence(55, 56). RNAss represents the stemness indices determined from expression data, whereas DNAss serves as the dryness index derived from methylation data. As the stemness index approaches 1, the degree of cell differentiation tends to decrease, while the characteristics of stem cells become more pronounced. Our analysis of SMAD3 expression and the tumor stemness markers DNAss and RNAss revealed significant negative correlations between SMAD3 expression and DNAss in four tumor types: COAD, COADREAD, BRCA, and UCEC. Conversely, In other tumor categories, there were evident and statistically significant positive correlations between SMAD3 expression and DNAss, including CESC, STES, THYM, THCA, UVM, and HNSC. In all tumors except PCPG, SMAD3 showed a significant inverse correlation with RNAss. These findings suggest that high tumor cell stemness is associated with low SMAD3 expression, which may promote tumor growth and metastasis. In the context of immune checkpoints, we identified over 30 immune checkpoints that exhibited positive correlations with most cancers, with the exception of KICH, ESCA, ALL, CHOL, and MESO. Additionally, our analysis indicated that low expression of SMAD3 often predicted a favorable response to immunotherapy. Collectively, these results demonstrate the close relationship between SMAD3, TMB, MSI, MMR, DNAss, RNAss, and immune checkpoints, highlighting the potential of SMAD3 as a novel biomarker for predicting the efficacy of immune checkpoint inhibitors (ICIs).
Although the contribution of SMAD3 to tumorigenesis is well-established, the precise underlying mechanism remains elusive. To elucidate SMAD3's biological functions in pancreatic adenocarcinoma (PAAD) and adrenocortical carcinoma (ACC), we conducted functional enrichment analysis. Our findings demonstrated that high SMAD3 expression was primarily associated with Interferon Gamma Response, Glycine, Serine, and Threonine Metabolism, as well as five signaling pathways, including the Notch Signaling Pathway, TGF-β Signaling Pathway, Wnt Signaling Pathway, Neuroactive Ligand Receptor Interaction, and P53 Pathway. The findings proffer an intriguing indication of the plausible implication of SMAD3 in the instigation of tumorigenesis and the intricate process of metastatic dissemination. Importantly, antecedent investigations into the realm of COVID-19 viral affliction have underscored the pivotal participation of SMAD3 in the pathogenesis of this malady, accentuating the conceivable therapeutic potency inherent in the abrogation or inhibition of SMAD3 as a prospective stratagem for disease amelioration (57). The prospect of SMAD3 serving as a focal point for anticancer interventions remains contingent upon a need for extended empirical inquiry. Furthermore, our investigation has unveiled potential therapeutic agents by scrutinizing the congruence between SMAD3 expression and drug sensitivity, thereby affirming SMAD3's standing as a viable candidate for tumor-directed therapies. Furthermore, a series of in vitro experiments conducted with hepatocellular carcinoma (LIHC) cells has illuminated the role of SMAD3 as an oncogenic entity, vigorously fostering the proliferation, migratory behavior, and invasive tendencies characteristic of LIHC cells. These findings, in a compelling manner, underscore the promotional influence of SMAD3 in orchestrating the proliferation and migratory capacities inherent to LIHC cells.
Several limitations are inherent to our study. First, there is a need for additional experimental confirmation, including immunohistochemistry and immunocytochemistry, to validate our findings. Second, the molecular regulatory mechanisms related to SMAD3 in various cancers require further exploration. Finally, it's essential to acknowledge the potential for systematic bias since we retrieved data from multiple databases for our analysis. These limitations underscore the need for more extensive research to fully elucidate SMAD3's role in cancer and its potential as a target for anticancer therapies.